Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Christina Ehrhardt is active.

Publication


Featured researches published by Christina Ehrhardt.


Nature Medicine | 2007

Mrp8 and Mrp14 are endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-induced shock

Thomas Vogl; Klaus Tenbrock; Stephan Ludwig; Nadja Leukert; Christina Ehrhardt; Marieke A. D. van Zoelen; Wolfgang Nacken; Dirk Foell; Tom van der Poll; Clemens Sorg; J. Roth

To identify new components that regulate the inflammatory cascade during sepsis, we characterized the functions of myeloid-related protein-8 (Mrp8, S100A8) and myeloid-related protein-14 (Mrp14, S100A9), two abundant cytoplasmic proteins of phagocytes. We now demonstrate that mice lacking Mrp8-Mrp14 complexes are protected from endotoxin-induced lethal shock and Escherichia coli–induced abdominal sepsis. Both proteins are released during activation of phagocytes, and Mrp8-Mrp14 complexes amplify the endotoxin-triggered inflammatory responses of phagocytes. Mrp8 is the active component that induces intracellular translocation of myeloid differentiation primary response protein 88 and activation of interleukin-1 receptor–associated kinase-1 and nuclear factor-κB, resulting in elevated expression of tumor necrosis factor-α (TNF-α). Using phagocytes expressing a nonfunctional Toll-like receptor 4 (TLR4), HEK293 cells transfected with TLR4, CD14 and MD2, and by surface plasmon resonance studies in vitro, we demonstrate that Mrp8 specifically interacts with the TLR4-MD2 complex, thus representing an endogenous ligand of TLR4. Therefore Mrp8-Mrp14 complexes are new inflammatory components that amplify phagocyte activation during sepsis upstream of TNFα–dependent effects.


Nature Cell Biology | 2001

Influenza virus propagation is impaired by inhibition of the Raf/MEK/ERK signalling cascade

Stephan Pleschka; Thorsten Wolff; Christina Ehrhardt; Gerd Hobom; Oliver Planz; Ulf R. Rapp; Stephan Ludwig

Influenza A viruses are important worldwide pathogens in humans and different animal species. The functions of most of the ten different viral proteins of this negative-strand RNA virus have been well elucidated. However, little is known about the virus-induced intracellular signalling events that support viral replication. The Raf/MEK/ERK cascade is the prototype of mitogen-activated protein (MAP) kinase cascades and has an important role in cell growth, differentiation and survival. Investigation of the function of this pathway has been facilitated by the identification of specific inhibitors such as U0126, which blocks the cascade at the level of MAPK/ERK kinase (MEK). Here we show that infection of cells with influenza A virus leads to biphasic activation of the Raf/MEK/ERK cascade. Inhibition of Raf signalling results in nuclear retention of viral ribonucleoprotein complexes (RNPs), impaired function of the nuclear-export protein (NEP/NS2) and concomitant inhibition of virus production. Thus, signalling through the mitogenic cascade seems to be essential for virus production and RNP export from the nucleus during the viral life cycle.


The FASEB Journal | 2003

IFN-α antagonistic activity of HCV core protein involves induction of suppressor of cytokine signaling-3

Johannes G. Bode; Stephan Ludwig; Christina Ehrhardt; Ute Albrecht; A. Erhardt; Fred Schaper; Peter C. Heinrich; Dieter Häussinger

Eighty percent of patients newly infected with the hepatitis C virus (HCV) develop chronic infection, suggesting that HCV can develop effective strategies to escape the unspecific and specific immune response of the host. Because SOCS molecules have been recognized to be powerful inhibitors of cytokine signaling via the Jak/STAT pathway, virus‐induced expression of these molecules should be an efficient instrument to counteract the cellular response toward interferons (IFNs), an essential part of first line antiviral immune response. This study shows that overexpression of HCV core protein inhibits IFN‐α‐induced tyrosine phosphorylation and activation of STAT1 in hepatic cells. With the use of a STAT1‐YFP fusion protein, further evidence is given that HCV core is capable to inhibit nuclear translocation of STAT1. Inhibition of STATl‐tyrosine phosphorylation was accompanied by the induction of SOCS3‐mRNA expression, suggesting that the HCV core protein impairs IFN‐α‐induced signal transduction via induction of SOCS3 expression. HCV core protein was competent to partially rescue growth of a genetically engineered influenza A virus lacking its own IFN antagonist. These IFN‐antagonistic properties of the HCV core protein may be part of the molecular basis of IFN‐a unresponsiveness in about one‐half of chronically infected HCV‐patients.


The EMBO Journal | 2003

Caspase 3 activation is essential for efficient influenza virus propagation

Walter J. Wurzer; Oliver Planz; Christina Ehrhardt; Martin Giner; Tobias Silberzahn; Stephan Pleschka; Stephan Ludwig

Apoptosis is a hallmark event observed upon infection with many viral pathogens, including influenza A virus. The apoptotic process is executed by a proteolytic system consisting of a family of cysteinyl proteases, termed caspases. Since the consequences of apoptosis induction and caspase activation for the outcome of an influenza virus infection are not clear, we have addressed this issue by interfering with expression or function of a major virus‐induced apoptosis effector, caspase 3. Surprisingly, influenza virus propagation was strongly impaired in the presence of an inhibitor that blocks caspase 3 and in cells where caspase 3 was partially knocked down by small interfering RNAs. Consistent with these findings, poor replication efficiencies of influenza A viruses in cells deficient for caspase 3 could be boosted 30‐fold by ectopic expression of the protein. Mechanistically, the block in virus propagation appeared to be due to retention of the viral RNP complexes in the nucleus, preventing formation of progeny virus particles. Our findings indicate that caspase 3 activation during the onset of apoptosis is a crucial event for efficient influenza virus propagation.


Journal of Virology | 2007

Influenza A Virus NS1 Protein Activates the PI3K/Akt Pathway To Mediate Antiapoptotic Signaling Responses

Christina Ehrhardt; Thorsten Wolff; Stephan Pleschka; Oliver Planz; Wiebke Beermann; Johannes G. Bode; Mirco Schmolke; Stephan Ludwig

ABSTRACT Recently we have shown that influenza A virus infection leads to activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway and that this cellular reaction is dependent on the expression of the viral nonstructural protein 1 (NS1). These data also suggested that PI3K activation confers a virus-supporting activity at intermediate stages of the infection cycle. So far it is not known which process is regulated by the kinase that supports virus replication. It is well established that upon infection with influenza A virus, the expression of the viral NS1 keeps the induction of beta interferon and the apoptotic response within a tolerable limit. On a molecular basis, this activity of NS1 has been suggested to preclude the activation of cellular double-stranded RNA receptors as well as impaired modulation of mRNA processing. Here we present a novel mode of action of the NS1 protein to suppress apoptosis induction. NS1 binds to and activates PI3K, which results in the activation of the PI3K effector Akt. This leads to a subsequent inhibition of caspase 9 and glycogen synthase-kinase 3β and limitation of the virus-induced cell death program. Thus, NS1 not only blocks but also activates signaling pathways to ensure efficient virus replication.


Journal of Virology | 2002

The Influenza A Virus NS1 Protein Inhibits Activation of Jun N-Terminal Kinase and AP-1 Transcription Factors

Stephan Ludwig; Xiuyan Wang; Christina Ehrhardt; Hongyong Zheng; Nicola R. Donelan; Oliver Planz; Stephan Pleschka; Adolfo García-Sastre; Gudrun Heins; Thorsten Wolff

ABSTRACT The influenza A virus nonstructural NS1 protein is known to modulate host cell gene expression and to inhibit double-stranded RNA (dsRNA)-mediated antiviral responses. Here we identify NS1 as the first viral protein that antagonizes virus- and dsRNA-induced activation of the stress response-signaling pathway mediated through Jun N-terminal kinase.


PLOS Pathogens | 2010

The Epidermal Growth Factor Receptor (EGFR) Promotes Uptake of Influenza A Viruses (IAV) into Host Cells

Thorsten Eierhoff; Eike R. Hrincius; Ursula Rescher; Stephan Ludwig; Christina Ehrhardt

Influenza A viruses (IAV) bind to sialic-acids at cellular surfaces and enter cells by using endocytotic routes. There is evidence that this process does not occur constitutively but requires induction of specific cellular signals, including activation of PI3K that promotes virus internalization. This implies engagement of cellular signaling receptors during viral entry. Here, we present first indications for an interplay of IAV with receptor tyrosine kinases (RTKs). As representative RTK family-members the epidermal growth factor receptor (EGFR) and the c-Met receptor were studied. Modulation of expression or activity of both RTKs resulted in altered uptake of IAV, showing that these receptors transmit entry relevant signals upon virus binding. More detailed studies on EGFR function revealed that virus binding lead to clustering of lipid-rafts, suggesting that multivalent binding of IAV to cells induces a signaling platform leading to activation of EGFR and other RTKs that in turn facilitates IAV uptake.


Cellular Microbiology | 2007

Acetylsalicylic acid (ASA) blocks influenza virus propagation via its NF-kappa B-inhibiting activity

Igor Mazur; Walter J. Wurzer; Christina Ehrhardt; Stephan Pleschka; Pilaipan Puthavathana; Tobias Silberzahn; Thorsten Wolff; Oliver Planz; Stephan Ludwig

Influenza is still one of the major plagues worldwide. The statistical likeliness of a new pandemic outbreak highlights the urgent need for new and amply available antiviral drugs. We and others have shown that influenza virus misuses the cellular IKK/NF‐κB signalling pathway for efficient replication suggesting that this module may be a suitable target for antiviral intervention. Here we examined acetylsalicylic acid (ASA), also known as aspirin, a widely used drug with a well‐known capacity to inhibit NF‐κB. We show that the drug efficiently blocks influenza virus replication in vitro and in vivo in a mechanism involving impaired expression of proapoptotic factors, subsequent inhibition of caspase activation as well as block of caspase‐mediated nuclear export of viral ribonucleoproteins. As ASA showed no toxic side‐effects or the tendency to induce resistant virus variants, existing salicylate‐based aerosolic drugs may be suitable as anti‐influenza agents. This is the first demonstration that specific targeting of a cellular factor is a suitable approach for anti‐influenza virus intervention.


FEBS Letters | 2004

Rac1 and PAK1 are upstream of IKK-ε and TBK-1 in the viral activation of interferon regulatory factor-3

Christina Ehrhardt; Christian Kardinal; Walter J. Wurzer; Thorsten Wolff; Christoph von Eichel-Streiber; Stephan Pleschka; Oliver Planz; Stephan Ludwig

The anti‐viral type I interferon (IFN) response is initiated by the immediate induction of IFNβ, which is mainly controlled by the IFN‐regulatory factor‐3 (IRF‐3). The signaling pathways mediating viral IRF‐3 activation are only poorly defined. We show that the Rho GTPase Rac1 is activated upon virus infection and controls IRF‐3 phosphorylation and activity. Inhibition of Rac1 leads to reduced IFNβ promoter activity and to enhanced virus production. As a downstream mediator of Rac signaling towards IRF‐3, we have identified the kinase p21‐activated kinase (PAK1). Furthermore, both Rac1 and PAK1 regulate the recently described IRF‐3 activators, IκB kinase‐ε and TANK‐binding kinase‐1, establishing a first canonical virus‐induced IRF‐3 activating pathway.


Cellular Microbiology | 2009

A new player in a deadly game: influenza viruses and the PI3K/Akt signalling pathway.

Christina Ehrhardt; Stephan Ludwig

Upon influenza A virus infection of cells, a wide variety of antiviral and virus‐supportive signalling pathways are induced. Phosphatidylinositol‐3‐kinase (PI3K) is a recent addition to the growing list of signalling mediators that are activated by these viruses. Several studies have addressed the role of PI3K and the downstream effector protein kinase Akt in influenza A virus‐infected cells. PI3K/Akt signalling is activated by diverse mechanisms in a biphasic manner and is required for multiple functions during infection. While the kinase supports activation of the interferon regulatory factor‐3 during antiviral interferon induction, it also exhibits virus supportive functions. In fact, PI3K not only regulates a very early step during viral entry but also results in suppression of premature apoptosis at later stages of infection. The latter function is dependent on the expression of the viral non‐structural protein‐1 (A/NS1). It has been shown that PI3K activation occurs by direct interaction of A/NS1 with the p85 regulatory subunit and interaction sites of A/NS1 and p85 have now been mapped in detail. Here, we summarize the current knowledge on influenza virus‐induced PI3K signalling and how this pathway supports viral propagation.

Collaboration


Dive into the Christina Ehrhardt's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Oliver Planz

Friedrich Loeffler Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karoline Droebner

Friedrich Loeffler Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge